1
|
Chung WC, Zhang S, Atfi A, Xu K. Lfng-expressing centroacinar cell is a unique cell-of-origin for p53 deficient pancreatic cancer. Oncogene 2024:10.1038/s41388-024-03226-7. [PMID: 39548190 DOI: 10.1038/s41388-024-03226-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies with limited understanding of etiology. Studies in mice showed that both acinar and ductal cells of the pancreas can be targeted by combination of oncogenic Kras and p53 mutations to form PDAC. How the transforming capacities of pancreatic cells are constrained, and whether a subset of cells could serve as a prime target for oncogenic transformation, remain obscure. Here we report that expression of a Notch modulator, Lunatic Fringe (Lfng), is restricted to a limited number of cells with centroacinar location and morphology in the adult pancreas. Lfng-expressing cells are preferentially targeted by oncogenic Kras along with p53 deletion to form PDAC, and deletion of Lfng blocks tumor initiation from these cells. Notch3 is a functional Notch receptor for PDAC initiation and progression in this context. Lfng is upregulated in acinar- and ductal-derived PDAC and its deletion suppresses these tumors. Finally, high LFNG expression is associated with high grade and poor survival in human patients. Taken together, Lfng marks a centroacinar subpopulation that is uniquely susceptible to oncogenic transformation when p53 is lost, and Lfng functions as an oncogene in all three lineages of the exocrine pancreas.
Collapse
Affiliation(s)
- Wen-Cheng Chung
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, 39216, USA
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, 39216, USA
| | - Shubing Zhang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 410013, China
| | - Azeddine Atfi
- Department of Biochemistry and Molecular Biology, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Keli Xu
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, 39216, USA.
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, 39216, USA.
| |
Collapse
|
2
|
Zhai W, Tian H, Liang X, Wu Y, Wen J, Liu Z, Zhao X, Tao L, Zou K. Androgen blockage impairs proliferation and function of Sertoli cells via Wee1 and Lfng. Cell Commun Signal 2024; 22:498. [PMID: 39407201 PMCID: PMC11481299 DOI: 10.1186/s12964-024-01875-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/05/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Androgens are essential hormones for testicular development and the maintenance of male fertility. Environmental factors, stress, aging, and psychological conditions can disrupt androgen production, impacting the androgen signaling pathway and consequently spermatogenesis. Within the testes, testosterone is produced by Leydig cells and acts on Sertoli cells by activating the androgen receptor (AR), which then translocates to the nucleus to function as a transcription factor. Despite clinical correlations between low testosterone levels and diminished sperm quality, the precise mechanism remains unclear. METHODS This study explores the hypothesis that reduced androgen levels impair Sertoli cell function by disrupting AR transcriptional regulation. Using an androgen blockade model with enzalutamide, we investigated the impact of low androgen levels on AR target genes in Sertoli cells through ChIP-seq and RNA-seq assays. RESULTS Our results reveal that androgen blockage increases AR enrichment on the promoter region of Wee1, promoting Wee1 expression, while decreasing binding to the promoter region of Lfng, inhibiting its expression. Increased WEE1 protein inhibits Sertoli cell proliferation, whereas reduced LFNG affects Notch modification, leading to decreased production of glial cell line-derived neurotrophic factor (GDNF), a key growth factor for spermatogonial stem cell self-renewal. CONCLUSIONS These findings provide new insights into the molecular mechanisms by which low androgen levels interfere with Sertoli cell functions, offering novel perspectives for the clinical treatment of male reproductive disorders.
Collapse
Affiliation(s)
- Wenhui Zhai
- Department of Emergency, The 305 Hospital of People's Liberation Army, Beijing, 100017, China
| | - Hairui Tian
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xuemei Liang
- General Surgery, The 305 Hospital of People's Liberation Army, Beijing, 100017, China
| | - Yunqiang Wu
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jian Wen
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, 210095, China
| | - Zhipeng Liu
- General Surgery, The 305 Hospital of People's Liberation Army, Beijing, 100017, China
| | - Xiaodong Zhao
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Li Tao
- Department of Emergency, The 305 Hospital of People's Liberation Army, Beijing, 100017, China.
| | - Kang Zou
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
3
|
Zhou K, Liu Y, Yuan S, Zhou Z, Ji P, Huang Q, Wen F, Li Q. Signalling in pancreatic cancer: from pathways to therapy. J Drug Target 2023; 31:1013-1026. [PMID: 37869884 DOI: 10.1080/1061186x.2023.2274806] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 10/18/2023] [Indexed: 10/24/2023]
Abstract
Pancreatic cancer (PC) is a common malignant tumour in the digestive system. Due to the lack of sensitive diagnostic markers, strong metastasis ability, and resistance to anti-cancer drugs, the prognosis of PC is inferior. In the past decades, increasing evidence has indicated that the development of PC is closely related to various signalling pathways. With the exploration of RAS-driven, epidermal growth factor receptor, Hedgehog, NF-κB, TGF-β, and NOTCH signalling pathways, breakthroughs have been made to explore the mechanism of pancreatic carcinogenesis, as well as the novel therapies. In this review, we discussed the signalling pathways involved in PC and summarised current targeted agents in the treatment of PC. Furthermore, opportunities and challenges in the exploration of potential therapies targeting signalling pathways were also highlighted.
Collapse
Affiliation(s)
- Kexun Zhou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yingping Liu
- The Second Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, China
| | | | - Ziyu Zhou
- The Second Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Pengfei Ji
- The Second Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Qianhan Huang
- School of Public Health, Xuzhou Medical University, Xuzhou, China
| | - Feng Wen
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qiu Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Gong X, Zheng C, Jia H, Liu Y, Yang R, Chen Z, Pan Y, Li X, Liu Y. A pan-cancer analysis revealing the role of LFNG, MFNG and RFNG in tumor prognosis and microenvironment. BMC Cancer 2023; 23:1065. [PMID: 37932706 PMCID: PMC10626706 DOI: 10.1186/s12885-023-11545-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 10/18/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND Fringe is a glycosyltransferase involved in tumor occurrence and metastasis. However, a comprehensive analysis of the Fringe family members lunatic fringe (LFNG), manic fringe (MFNG), radical fringe (RFNG) in human cancers is lacking. METHODS In this study, we performed a pan-cancer analysis of Fringe family members in 33 cancer types with transcriptomic, genomic, methylation data from The Cancer Genome Atlas (TCGA) project. The correlation between Fringe family member expression and patient overall survival, copy number variation, methylation, Gene Ontology enrichment, and tumor-infiltrating lymphocytes (TILs) was investigated by using multiple databases, such as cBioPortal, Human Protein Atlas, GeneCards, STRING, MSigDB, TISIDB, and TIMER2. In vitro experiments and immunohistochemical assays were performed to validate our findings. RESULTS High expression levels of LFNG, MFNG, RFNG were closely associated with poor overall survival in multiple cancers, particularly in pancreatic adenocarcinoma (PAAD), uveal melanoma (UVM), and brain lower-grade glioma (LGG). Copy number variation analysis revealed that diploid and gain mutations of LFNG was significantly increased in PAAD and stomach adenocarcinoma (STAD), and significantly associated with the methylation levels in promoter regions. Significant differential genes between high and low expression groups of these Fringe family members were found to be consistently enriched in immune response and T cell activation pathway, extracellular matrix adhesion pathway, RNA splicing and ion transport pathways. Correlation between the abundance of tumor-infiltrating lymphocytes (TILs) and LFNG, MFNG, and RFNG expression showed that high LFNG expression was associated with lower TIL levels, particularly in PAAD. In vitro experiment by using pancreatic cancer PANC1 cells showed that LFNG overexpression promoted cell proliferation and invasion. Immunohistochemical assay in 90 PAAD patients verified the expression level of LFNG and its relationship with the prognosis. CONCLUSIONS Our study provides a relatively comprehensive understanding of the expression, mutation, copy number, promoter methylation level changes along with prognosis values of LFNG, MFNG, and RFNG in different tumors. High LFNG expression may serve as a poor prognosis molecular marker for PAAD.
Collapse
Affiliation(s)
- Xun Gong
- Department of Hepatobiliary Surgery, Shenzhen Key Laboratory, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University, 1098 Xueyuan Avenue, Nanshan District, Shenzhen, 518000, Guangdong, P.R. China
| | - Chenglong Zheng
- Department of Hepatobiliary Surgery, Shenzhen Key Laboratory, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University, 1098 Xueyuan Avenue, Nanshan District, Shenzhen, 518000, Guangdong, P.R. China
| | - Haiying Jia
- Department of Hepatobiliary Surgery, Shenzhen Key Laboratory, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University, 1098 Xueyuan Avenue, Nanshan District, Shenzhen, 518000, Guangdong, P.R. China
| | - Yangruiyu Liu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Rui Yang
- Department of Hepatobiliary Surgery, Shenzhen Key Laboratory, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University, 1098 Xueyuan Avenue, Nanshan District, Shenzhen, 518000, Guangdong, P.R. China
| | - Zizhou Chen
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Yihang Pan
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China.
- Big Data Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China.
| | - Xiaowu Li
- Department of Hepatobiliary Surgery, Shenzhen Key Laboratory, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University, 1098 Xueyuan Avenue, Nanshan District, Shenzhen, 518000, Guangdong, P.R. China.
| | - Yuchen Liu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China.
- Big Data Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China.
| |
Collapse
|
5
|
Qian Q, Song J, Pu Q, Chen C, Yan J, Wang H. Acute/chronic exposure to bisphenol A induced immunotoxicity in zebrafish and its potential association with pancreatic cancer risk. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 258:106514. [PMID: 37019016 DOI: 10.1016/j.aquatox.2023.106514] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 06/19/2023]
Abstract
Previous studies have confirmed that bisphenol A (BPA) induced immune toxicity and affected diseases, however, the underlying mechanism remains unknown. In the present study, zebrafish was employed as the model to assess the immunotoxicity and the potential disease risk of BPA exposure. Upon BPA exposure, a series of abnormalities were found, which included the increased oxidative stress, damaged innate and adaptive immune functions and the elevated insulin and blood glucose levels. According to the target prediction and RNA sequencing data of BPA, the differential expression genes were found enriched in immune- and pancreatic cancer-related pathway and process, and the potential role of stat3 in the regulation of these processes was revealed. The key immune- and pancreatic cancer-related genes were selected for further confirmation by RT-qPCR. Based on the changes in the expression levels of these genes, our hypothesis that BPA induced the occurrence of pancreatic cancer by modulating immune responses was further evidenced. Deeper mechanism was further disclosed by molecular dock simulation and survival analysis of key genes, proving that BPA stably bound to STAT3 and IL10 and STAT3 may serve as the target of BPA-inducing pancreatic cancer. These results are of great significance in deepening the molecular mechanism of immunotoxicity induced by BPA and our understanding of the risk assessment of contaminants.
Collapse
Affiliation(s)
- Qiuhui Qian
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, PR. China
| | - Jie Song
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, PR. China
| | - Qian Pu
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, PR. China
| | - Chen Chen
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, PR. China
| | - Jin Yan
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, PR. China
| | - Huili Wang
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, PR. China.
| |
Collapse
|
6
|
Chung WC, Xu K. Notch signaling pathway in pancreatic tumorigenesis. Adv Cancer Res 2023. [DOI: 10.1016/bs.acr.2023.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
|
7
|
Mugisha S, Di X, Disoma C, Jiang H, Zhang S. Fringe family genes and their modulation of Notch signaling in cancer. Biochim Biophys Acta Rev Cancer 2022; 1877:188746. [PMID: 35660646 DOI: 10.1016/j.bbcan.2022.188746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 05/30/2022] [Accepted: 05/30/2022] [Indexed: 11/26/2022]
Abstract
Fringes are glycosyltransferases that transfer N-acetylglucosamine to the O-linked fucose of Notch receptors. They regulate the Notch signaling activity that drives tumor formation and progression, resulting in poor prognosis. However, the specific tumor-promoting role of Fringes differs depending on the type of cancer. Although a particular Fringe member could act as a tumor suppressor in one cancer type, it may act as an oncogene in another. This review discusses the tumorigenic role of the Fringe family (lunatic fringe, manic fringe, and radical fringe) in modulating Notch signaling in various cancers. Although the crucial functions of Fringes continue to emerge as more mechanistic studies are being pursued, further translational research is needed to explore their roles and therapeutic benefits in various malignancies.
Collapse
Affiliation(s)
- Samson Mugisha
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, PR China
| | - Xiaotang Di
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, PR China
| | - Cyrollah Disoma
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, PR China
| | - Hao Jiang
- Department of Biomedical Informatics, School of Life Sciences, Central South University, Changsha, Hunan 410013, PR China.
| | - Shubing Zhang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, PR China; Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, Hunan 410013, PR China.
| |
Collapse
|
8
|
Wang W, Okajima T, Takeuchi H. Significant Roles of Notch O-Glycosylation in Cancer. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27061783. [PMID: 35335147 PMCID: PMC8950332 DOI: 10.3390/molecules27061783] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/05/2022] [Accepted: 03/07/2022] [Indexed: 12/27/2022]
Abstract
Notch signaling, which was initially identified in Drosophila wing morphogenesis, plays pivotal roles in cell development and differentiation. Optimal Notch pathway activity is essential for normal development and dysregulation of Notch signaling leads to various human diseases, including many types of cancers. In hematopoietic cancers, such as T-cell acute lymphoblastic leukemia, Notch plays an oncogenic role, while in acute myeloid leukemia, it has a tumor-suppressive role. In solid tumors, such as hepatocellular carcinoma and medulloblastoma, Notch may have either an oncogenic or tumor-suppressive role, depending on the context. Aberrant expression of Notch receptors or ligands can alter the ligand-dependent Notch signaling and changes in trafficking can lead to ligand-independent signaling. Defects in any of the two signaling pathways can lead to tumorigenesis and tumor progression. Strikingly, O-glycosylation is one such process that modulates ligand–receptor binding and trafficking. Three types of O-linked modifications on the extracellular epidermal growth factor-like (EGF) repeats of Notch receptors are observed, namely O-glucosylation, O-fucosylation, and O-N-acetylglucosamine (GlcNAc) modifications. In addition, O-GalNAc mucin-type O-glycosylation outside the EGF repeats also appears to occur in Notch receptors. In this review, we first briefly summarize the basics of Notch signaling, describe the latest information on O-glycosylation of Notch receptors classified on a structural basis, and finally describe the regulation of Notch signaling by O-glycosylation in cancer.
Collapse
Affiliation(s)
- Weiwei Wang
- Department of Molecular Biochemistry, Nagoya University School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan; (W.W.); (T.O.)
| | - Tetsuya Okajima
- Department of Molecular Biochemistry, Nagoya University School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan; (W.W.); (T.O.)
- Institute for Glyco-Core Research (iGCORE), Integrated Glyco-Biomedical Research Center, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Hideyuki Takeuchi
- Department of Molecular Biochemistry, Nagoya University School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan; (W.W.); (T.O.)
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
- Correspondence:
| |
Collapse
|
9
|
Chen WK, Oon CE, Kaur G, Sainson RC, Li JL. Downregulation of Manic fringe impedes angiogenesis and cell migration of renal carcinoma. Microvasc Res 2022; 142:104341. [DOI: 10.1016/j.mvr.2022.104341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/14/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022]
|
10
|
Mohamed Abd-El-Halim Y, El Kaoutari A, Silvy F, Rubis M, Bigonnet M, Roques J, Cros J, Nicolle R, Iovanna J, Dusetti N, Mas E. A glycosyltransferase gene signature to detect pancreatic ductal adenocarcinoma patients with poor prognosis. EBioMedicine 2021; 71:103541. [PMID: 34425307 PMCID: PMC8379629 DOI: 10.1016/j.ebiom.2021.103541] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 07/26/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is characterized by an important heterogeneity, reflected by different clinical outcomes and chemoresistance. During carcinogenesis, tumor cells display aberrant glycosylated structures, synthetized by deregulated glycosyltransferases, supporting the tumor progression. In this study, we aimed to determine whether PDAC could be stratified through their glycosyltransferase expression profiles better than the current binary classification (basal-like and classical) in order to improve detection of patients with poor prognosis. Methods Bioinformatic analysis of 169 glycosyltransferase RNA sequencing data were performed for 74 patient-derived xenografts (PDX) of resected and unresectable tumors. The Australian cohort of International Cancer Genome Consortium and the microarray dataset from Puleo patient's cohort were used as independent validation datasets. Findings New PDAC stratification based on glycosyltransferase expression profile allowed to distinguish different groups of patients with distinct clinical outcome (p-value = 0.007). A combination of 19 glycosyltransferases differentially expressed in PDX defined a glyco-signature, whose prognostic value was validated on datasets including resected whole tumor tissues. The glyco-signature was able to discriminate three clusters of PDAC patients on the validation cohorts, two clusters displaying a short overall survival compared to one cluster having a better prognosis. Both poor prognostic clusters having different glyco-profiles in Puleo patient's cohort were correlated with stroma activated or desmoplastic subtypes corresponding to distinct microenvironment features (p-value < 0.0001). Besides, differential expression and enrichment analyses revealed deregulated functional pathways specific to different clusters. Interpretation This study identifies a glyco-signature relevant for a prognostic use, potentially applicable to resected and unresectable PDAC. Furthermore, it provides new potential therapeutic targets. Funding This work was supported by INCa (Grants number 2018-078 and 2018-079), Fondation ARC (Grant number ARCPJA32020070002326), Cancéropôle PACA, DGOS (labelization SIRIC, Grant number 6038), Amidex Foundation and Ligue Nationale Contre le Cancer and by institutional fundings from INSERM and the Aix-Marseille Université.
Collapse
Affiliation(s)
- Yousra Mohamed Abd-El-Halim
- Cancer Research Center of Marseille, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Abdessamad El Kaoutari
- Cancer Research Center of Marseille, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Françoise Silvy
- Cancer Research Center of Marseille, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Marion Rubis
- Cancer Research Center of Marseille, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Martin Bigonnet
- Cancer Research Center of Marseille, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Julie Roques
- Cancer Research Center of Marseille, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Jérôme Cros
- Department of Pathology, Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, France
| | - Rémy Nicolle
- Programme Cartes d'Identité des Tumeurs (CIT), Ligue Nationale Contre Le Cancer, Paris, France
| | - Juan Iovanna
- Cancer Research Center of Marseille, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Nelson Dusetti
- Cancer Research Center of Marseille, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France.
| | - Eric Mas
- Cancer Research Center of Marseille, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France.
| |
Collapse
|
11
|
Barua R, Mizuno K, Tashima Y, Ogawa M, Takeuchi H, Taguchi A, Okajima T. Bioinformatics and Functional Analyses Implicate Potential Roles for EOGT and L-fringe in Pancreatic Cancers. Molecules 2021; 26:molecules26040882. [PMID: 33562410 PMCID: PMC7915272 DOI: 10.3390/molecules26040882] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/30/2021] [Accepted: 02/02/2021] [Indexed: 12/14/2022] Open
Abstract
Notch signaling receptors, ligands, and their downstream target genes are dysregulated in pancreatic ductal adenocarcinoma (PDAC), suggesting a role of Notch signaling in pancreatic tumor development and progression. However, dysregulation of Notch signaling by post-translational modification of Notch receptors remains poorly understood. Here, we analyzed the Notch-modifying glycosyltransferase involved in the regulation of the ligand-dependent Notch signaling pathway. Bioinformatic analysis revealed that the expression of epidermal growth factor (EGF) domain-specific O-linked N-acetylglucosamine (EOGT) and Lunatic fringe (LFNG) positively correlates with a subset of Notch signaling genes in PDAC. The lack of EOGT or LFNG expression inhibited the proliferation and migration of Panc-1 cells, as observed by the inhibition of Notch activation. EOGT expression is significantly increased in the basal subtype, and low expression of both EOGT and LFNG predicts better overall survival in PDAC patients. These results imply potential roles for EOGT- and LFNG-dependent Notch signaling in PDAC.
Collapse
Affiliation(s)
- Rashu Barua
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan; (R.B.); (Y.T.); (M.O.); (H.T.)
| | - Kazuyuki Mizuno
- Division of Molecular Diagnostics, Aichi Cancer Center, 1-1 Kanokoden, Chikusa-ku, Nagoya, Aichi 464-8681, Japan; (K.M.); (A.T.)
| | - Yuko Tashima
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan; (R.B.); (Y.T.); (M.O.); (H.T.)
- Institute for Glyco-core Research (iGCORE), Integrated Glyco-Biomedical Research Center, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601 Nagoya, Japan
| | - Mitsutaka Ogawa
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan; (R.B.); (Y.T.); (M.O.); (H.T.)
- Institute for Glyco-core Research (iGCORE), Integrated Glyco-Biomedical Research Center, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601 Nagoya, Japan
| | - Hideyuki Takeuchi
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan; (R.B.); (Y.T.); (M.O.); (H.T.)
- Institute for Glyco-core Research (iGCORE), Integrated Glyco-Biomedical Research Center, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601 Nagoya, Japan
| | - Ayumu Taguchi
- Division of Molecular Diagnostics, Aichi Cancer Center, 1-1 Kanokoden, Chikusa-ku, Nagoya, Aichi 464-8681, Japan; (K.M.); (A.T.)
- Division of Advanced Cancer Diagnostics, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan
| | - Tetsuya Okajima
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan; (R.B.); (Y.T.); (M.O.); (H.T.)
- Institute for Glyco-core Research (iGCORE), Integrated Glyco-Biomedical Research Center, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601 Nagoya, Japan
- Correspondence: ; Tel.: +81-52-744-2068; Fax: +81-52-744-2069
| |
Collapse
|
12
|
Orzechowska M, Anusewicz D, Bednarek AK. Functional Gene Expression Differentiation of the Notch Signaling Pathway in Female Reproductive Tract Tissues-A Comprehensive Review With Analysis. Front Cell Dev Biol 2021; 8:592616. [PMID: 33384996 PMCID: PMC7770115 DOI: 10.3389/fcell.2020.592616] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022] Open
Abstract
The Notch pathway involves evolutionarily conserved signaling regulating the development of the female tract organs such as breast, ovary, cervix, and uterine endometrium. A great number of studies revealed Notch aberrancies in association with their carcinogenesis and disease progression, the management of which is still challenging. The present study is a comprehensive review of the available literature on Notch signaling during the normal development and carcinogenesis of the female tract organs. The review has been enriched with our analyses of the TCGA data including breast, cervical, ovarian, and endometrial carcinomas concerning the effects of Notch signaling at two levels: the core components and downstream effectors, hence filling the lack of global overview of Notch-driven carcinogenesis and disease progression. Phenotype heterogeneity regarding Notch signaling was projected in two uniform manifold approximation and projection algorithm dimensions, preceded by the principal component analysis step reducing the data burden. Additionally, overall and disease-free survival analyses were performed with the optimal cutpoint determination by Evaluate Cutpoints software to establish the character of particular Notch components in tumorigenesis. In addition to the review, we demonstrated separate models of the examined cancers of the Notch pathway and its targets, although expression profiles of all normal tissues were much more similar to each other than to its cancerous compartments. Such Notch-driven cancerous differentiation resulted in a case of opposite association with DFS and OS. As a consequence, target genes also show very distinct profiles including genes associated with cell proliferation and differentiation, energy metabolism, or the EMT. In conclusion, the observed Notch associations with the female tract malignancies resulted from differential expression of target genes. This may influence a future analysis to search for new therapeutic targets based on specific Notch pathway profiles.
Collapse
Affiliation(s)
| | - Dorota Anusewicz
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| | - Andrzej K Bednarek
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
13
|
Matsumoto K, Luther KB, Haltiwanger RS. Diseases related to Notch glycosylation. Mol Aspects Med 2020; 79:100938. [PMID: 33341260 DOI: 10.1016/j.mam.2020.100938] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/30/2020] [Accepted: 12/03/2020] [Indexed: 12/15/2022]
Abstract
The Notch receptors are a family of transmembrane proteins that mediate direct cell-cell interactions and control numerous cell-fate specifications in humans. The extracellular domains of mammalian Notch proteins contain 29-36 tandem epidermal growth factor-like (EGF) repeats, most of which have O-linked glycan modifications: O-glucose added by POGLUT1, O-fucose added by POFUT1 and elongated by Fringe enzymes, and O-GlcNAc added by EOGT. The extracellular domain is also N-glycosylated. Mutations in the glycosyltransferases modifying Notch have been identified in several diseases, including Dowling-Degos Disease (haploinsufficiency of POFUT1 or POGLUT1), a form of limb-girdle muscular dystrophy (autosomal recessive mutations in POGLUT1), Spondylocostal Dysostosis 3 (autosomal recessive mutations in LFNG), Adams-Oliver syndrome (autosomal recessive mutations in EOGT), and some cancers (amplification, gain or loss-of-function of POFUT1, Fringe enzymes, POGLUT1, MGAT3). Here we review the characteristics of these diseases and potential molecular mechanisms.
Collapse
Affiliation(s)
- Kenjiroo Matsumoto
- Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, 315 Riverbend Road, Athens, GA, 30602, USA
| | - Kelvin B Luther
- Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, 315 Riverbend Road, Athens, GA, 30602, USA
| | - Robert S Haltiwanger
- Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, 315 Riverbend Road, Athens, GA, 30602, USA.
| |
Collapse
|
14
|
Chung WC, Challagundla L, Zhou Y, Li M, Atfi A, Xu K. Loss of Jag1 cooperates with oncogenic Kras to induce pancreatic cystic neoplasms. Life Sci Alliance 2020; 4:4/2/e201900503. [PMID: 33268505 PMCID: PMC7756968 DOI: 10.26508/lsa.201900503] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 01/09/2023] Open
Abstract
Notch signaling exerts both oncogenic and tumor-suppressive functions in the pancreas. In this study, deletion of Jag1 in conjunction with oncogenic Kras G12D expression in the mouse pancreas induced rapid development of acinar-to-ductal metaplasia and early stage pancreatic intraepithelial neoplasm; however, culminating in cystic neoplasms rather than ductal adenocarcinoma. Most cystic lesions in these mice were reminiscent of serous cystic neoplasm, and the rest resembled intraductal papillary mucinous neoplasm. Jag1 expression was lost or decreased in cystic lesions but retained in adenocarcinoma in these mice, so was the expression of Sox9. In pancreatic cancer patients, JAG1 expression is higher in cancerous tissue, and high JAG1 is associated with poor overall survival. Expression of SOX9 is correlated with JAG1, and high SOX9 is also associated with poor survival. Mechanistically, Jag1 regulates expression of Lkb1, a tumor suppressor involved in the development of pancreatic cystic neoplasm. Collectively, Jag1 can act as a tumor suppressor in the pancreas by delaying precursor lesions, whereas loss of Jag1 promoted a phenotypic switch from malignant carcinoma to benign cystic lesions.
Collapse
Affiliation(s)
- Wen-Cheng Chung
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - Lavanya Challagundla
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yunyun Zhou
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS, USA
| | - Min Li
- Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Azeddine Atfi
- Cellular and Molecular Pathogenesis Division, Department of Pathology, Virginia Commonwealth University, Richmond, VA, USA
| | - Keli Xu
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, USA .,Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
15
|
Meisel CT, Porcheri C, Mitsiadis TA. Cancer Stem Cells, Quo Vadis? The Notch Signaling Pathway in Tumor Initiation and Progression. Cells 2020; 9:cells9081879. [PMID: 32796631 PMCID: PMC7463613 DOI: 10.3390/cells9081879] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/04/2020] [Accepted: 08/05/2020] [Indexed: 02/06/2023] Open
Abstract
The Notch signaling pathway regulates cell proliferation, cytodifferentiation and cell fate decisions in both embryonic and adult life. Several aspects of stem cell maintenance are dependent from the functionality and fine tuning of the Notch pathway. In cancer, Notch is specifically involved in preserving self-renewal and amplification of cancer stem cells, supporting the formation, spread and recurrence of the tumor. As the function of Notch signaling is context dependent, we here provide an overview of its activity in a variety of tumors, focusing mostly on its role in the maintenance of the undifferentiated subset of cancer cells. Finally, we analyze the potential of molecules of the Notch pathway as diagnostic and therapeutic tools against the various cancers.
Collapse
|
16
|
Urata Y, Takeuchi H. Effects of Notch glycosylation on health and diseases. Dev Growth Differ 2019; 62:35-48. [PMID: 31886522 DOI: 10.1111/dgd.12643] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/03/2019] [Accepted: 12/03/2019] [Indexed: 12/13/2022]
Abstract
Notch signaling is an evolutionarily conserved signaling pathway and is essential for cell-fate specification in metazoans. Dysregulation of Notch signaling results in various human diseases, including cardiovascular defects and cancer. In 2000, Fringe, a known regulator of Notch signaling, was discovered as a Notch-modifying glycosyltransferase. Since then, glycosylation-a post-translational modification involving literal sugars-on the Notch extracellular domain has been noted as a critical mechanism for the regulation of Notch signaling. Additionally, the presence of diverse O-glycans decorating Notch receptors has been revealed in the extracellular domain epidermal growth factor-like (EGF) repeats. Here, we concisely summarize the recent studies in the human diseases associated with aberrant Notch glycosylation.
Collapse
Affiliation(s)
- Yusuke Urata
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hideyuki Takeuchi
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
17
|
Wang X, Wang R, Bai S, Xiong S, Li Y, Liu M, Zhao Z, Wang Y, Zhao Y, Chen W, Billiar TR, Cheng B. Musashi2 contributes to the maintenance of CD44v6+ liver cancer stem cells via notch1 signaling pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:505. [PMID: 31888685 PMCID: PMC6936093 DOI: 10.1186/s13046-019-1508-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 12/16/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Liver cancer stem cells (LCSCs) contribute to hepatocellular carcinoma (HCC) development, metastasis, and drug resistance. MSI2 and Notch1 signaling are involved in the maintenance of CSCs. However, it is unknown whether MSI2 and Notch1 are involved in the maintenance of CD44v6+ LCSCs. Therefore, we investigated the clinical significance and function of MSI2 and its relationship with Notch1 signaling in the maintenance of stemness properties in CD44v6+ LCSCs. METHODS The expression of MSI2 and CD44v6 were detected by fresh specimens and a HCC tissue microarray. The tissue microarray containing 82 HCC samples was used to analyze the correlation between CD44v6 and MSI2. CD44v6+/- cells were isolated using microbeads sorting. We explored the roles of MSI2 and Notch1 signaling in CD44v6+ LCSCs by sphere formation assay, transwell assay, clone formation assay in vitro, and xenograft tumor models in vivo. A Notch RT2 PCR Array, Co-immunoprecipitation, and RNA-immunoprecipitation were used to further investigate the molecular mechanism of MSI2 in activating Notch1 signaling. RESULTS Here, we found MSI2 expression was positively correlated with high CD44v6 expression in HCC tissues, and further correlated with tumor differentiation. CD44v6+ cells isolated from HCC cell lines exhibited increased self-renewal, proliferation, migration and invasion, resistance to Sorafenib and tumorigenic capacity. Both MSI2 and Notch1 signaling were elevated in sorted CD44v6+ cells than CD44v6- cells and played essential roles in the maintenance of stemness of CD44v6+ LCSCs. Mechanically, MSI2 directly bound to Lunatic fringe (LFNG) mRNA and protein, resulting in Notch1 activation. CONCLUSIONS Our results demonstrated that MSI2 maintained the stemness of CD44v6+ LCSCs by activating Notch1 signaling through the interaction with LFNG, which could be a potential molecular target for stem cell-targeted therapy for liver cancer.
Collapse
Affiliation(s)
- Xiju Wang
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China, 430030
| | - Ronghua Wang
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China, 430030
| | - Shuya Bai
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China, 430030
| | - Si Xiong
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China, 430030
| | - Yawen Li
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China, 430030
| | - Man Liu
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China, 430030
| | - Zhenxiong Zhao
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China, 430030
| | - Yun Wang
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China, 430030
| | - Yuchong Zhao
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China, 430030
| | - Wei Chen
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China, 430030
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Bin Cheng
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China, 430030.
| |
Collapse
|
18
|
Downregulation of Notch Signaling in Kras-Induced Gastric Metaplasia. Neoplasia 2019; 21:810-821. [PMID: 31276933 PMCID: PMC6611983 DOI: 10.1016/j.neo.2019.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/28/2019] [Accepted: 06/04/2019] [Indexed: 11/21/2022] Open
Abstract
Activating mutations and amplification of Kras and, more frequently, signatures for Kras activation are noted in stomach cancer. Expression of mutant KrasG12D in the mouse gastric mucosa has been shown to induce hyperplasia and metaplasia. However, the mechanisms by which Kras activation leads to gastric metaplasia are not fully understood. Here we report that KrasLSL-G12D/+;Pdx1-cre, a mouse model known for pancreatic cancer, also mediates KrasG12D expression in the stomach, causing gastric hyperplasia and metaplasia prior to the pathologic changes in the pancreas. These mice exhibit ectopic cell proliferation at the base of gastric glands, whereas wild-type mice contain proliferating cells primarily at the isthmus/neck of the gastric glands. Notch signaling is decreased in the KrasLSL-G12D/+;Pdx1-cre gastric mucosa, as shown by lower levels of cleaved Notch intracellular domains and downregulation of Notch downstream target genes. Expression of a Notch ligand Jagged1 is downregulated at the base of the mutant gland, accompanied by loss of chief cell marker Mist1. We demonstrate that exogenous Jagged1 or overexpression of Notch intracellular domain stimulates Mist1 expression in gastric cancer cell lines, suggesting positive regulation of Mist1 by Notch signaling. Finally, deletion of Jagged1 or Notch3 in KrasLSL-G12D/+;Pdx1-cre mice promoted development of squamous cell carcinoma in the forestomach, albeit short of invasive adenocarcinoma in the glandular stomach. Taken together, these results reveal downregulation of Notch signaling and Mist1 expression during the initiation of Kras-driven gastric tumorigenesis and suggest a tumor-suppressive role for Notch in this context.
Collapse
|
19
|
Lu X, Gao C, Liu C, Zhuang J, Su P, Li H, Wang X, Sun C. Identification of the key pathways and genes involved in HER2-positive breast cancer with brain metastasis. Pathol Res Pract 2019; 215:152475. [PMID: 31178227 DOI: 10.1016/j.prp.2019.152475] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/14/2019] [Accepted: 05/25/2019] [Indexed: 01/19/2023]
Abstract
BACKGROUND The risk of brain metastasis (BM) in HER2-positive (+) breast cancer (BC) patients is significantly higher than that in HER2-negative (-) BC patients. The high incidence and mortality rate makes it urgent to elucidate the key pathways and genes involved and identify patients who are more at risk of developing BM. MATERIALS AND METHODS To identify the target genes in HER2+BC patients with BM, we analyzed the microarray datasets (GSE43837) derived from the Gene Expression Omnibus (GEO) database. The GEO2R tool was used to extract the differentially expressed genes (DEGs) involved in HER2+ primary BC and BC with BM. Bioinformatics methods including Gene Ontology (GO) functional annotation analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed with the screened DEGs. The protein-protein interactions of the DEGs were analyzed using the Search Tool for the Retrieval of Interacting Genes (STRING) database and visualized using Cytoscape software. Finally, GSEA analysis was performed to identify the hub genes and the important pathways. RESULTS A total of 751 upregulated and 285 downregulated DEGs were identified. The GO function and KEGG pathway enrichment analyses indicated that the DEGs were all enriched in the protein binding molecular function. The top five hub nodes were screened out, included PHLPP1, UBC, ACACB, TGFB1, and ACTB. The GSEA results demonstrated that the five hub genes are mainly enriched in the ribosomal pathway. CONCLUSION Our study suggests that the five hub genes (PHLPP1, UBC, ACACB, TGFB1, and ACTB) are associated with HER2+BC with BM. The GSEA analysis revealed that the ribosomal pathway seems to play a very important role in the pathogenesis of HER2+BC with BM.
Collapse
Affiliation(s)
- Xin Lu
- Clinical Medical Colleges, Shandong University, Jinan 250100, Shandong, China; Department of Oncology, Taian Tumor Hospital, Taian 271000, Shandong, China
| | - Chundi Gao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong, China
| | - Cun Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong, China
| | - Jing Zhuang
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang 261041, Shandong, China
| | - Peiying Su
- Department of Oncology, Taian Tumor Hospital, Taian 271000, Shandong, China
| | - Huayao Li
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong, China
| | - Xue Wang
- Clinical Medical Colleges, Qingdao University, Qingdao 266071, Shandong, China
| | - Changgang Sun
- Department of Oncology, Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong, China; Department of Oncology, Affiliated hospital of Shandong University of Traditional Chinese Medicine, Weifang 261031, Shandong, China.
| |
Collapse
|
20
|
Holdener BC, Haltiwanger RS. Protein O-fucosylation: structure and function. Curr Opin Struct Biol 2019; 56:78-86. [PMID: 30690220 DOI: 10.1016/j.sbi.2018.12.005] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 12/18/2018] [Accepted: 12/19/2018] [Indexed: 12/22/2022]
Abstract
Fucose is a common terminal modification on protein and lipid glycans. Fucose can also be directly linked to protein via an O-linkage to Serine or Threonine residues located within consensus sequences contained in Epidermal Growth Factor-like (EGF) repeats and Thrombospondin Type 1 Repeats (TSRs). In this context, fucose is added exclusively to properly folded EGF repeats and TSRs by Protein O-fucosyltransferases 1 and 2, respectively. In both cases, the O-linked fucose can also be elongated with other sugars. Here, we describe the biological importance of these O-fucose glycans and molecular mechanisms by which they affect the function of the proteins they modify. O-Fucosylation of EGF repeats modulates the Notch signaling pathway, while O-fucosylation of TSRs is predicted to influence secretion of targets including several extracellular proteases. Recent data show O-fucose glycans mediate their effects by participating in both intermolecular and intramolecular interactions.
Collapse
Affiliation(s)
- Bernadette C Holdener
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | | |
Collapse
|
21
|
Varshney S, Stanley P. Multiple roles for O-glycans in Notch signalling. FEBS Lett 2018; 592:3819-3834. [PMID: 30207383 DOI: 10.1002/1873-3468.13251] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/05/2018] [Accepted: 09/06/2018] [Indexed: 12/12/2022]
Abstract
Notch signalling regulates a plethora of developmental processes and is also essential for the maintenance of tissue homeostasis in adults. Therefore, fine-tuning of Notch signalling strength needs to be tightly regulated. Of key importance for the regulation of Notch signalling are O-fucose, O-GlcNAc and O-glucose glycans attached to the extracellular domain of Notch receptors. The EGF repeats of the Notch receptor extracellular domain harbour consensus sites for addition of the different types of O-glycan to Ser or Thr, which takes place in the endoplasmic reticulum. Studies from Drosophila to mammals have demonstrated the multifaceted roles of O-glycosylation in regulating Notch signalling. O-glycosylation modulates different aspects of Notch signalling including recognition by Notch ligands, the strength of ligand binding, Notch receptor trafficking, stability and activation at the cell surface. Defects in O-glycosylation of Notch receptors give rise to pathologies in humans. This Review summarizes the nature of the O-glycans on Notch receptors and their differential effects on Notch signalling.
Collapse
Affiliation(s)
- Shweta Varshney
- Department of Cell Biology, Albert Einstein College of Medicine, New York, NY, USA
| | - Pamela Stanley
- Department of Cell Biology, Albert Einstein College of Medicine, New York, NY, USA
| |
Collapse
|
22
|
Giaimo BD, Borggrefe T. Introduction to Molecular Mechanisms in Notch Signal Transduction and Disease Pathogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1066:3-30. [DOI: 10.1007/978-3-319-89512-3_1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
23
|
Huang X, Zhi X, Gao Y, Ta N, Jiang H, Zheng J. LncRNAs in pancreatic cancer. Oncotarget 2018; 7:57379-57390. [PMID: 27429196 PMCID: PMC5302996 DOI: 10.18632/oncotarget.10545] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 06/17/2016] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer (PC) is one of the most common causes of cancer-related death. The underlying mechanism of PC is not completely understood at present. Studies in recent years have demonstrated that long non-coding RNAs (lncRNAs) have multiple biological functions in cell growth, differentiation and proliferation. Notably, expressions of some lncRNAs undergo significant changes in the initiation and progression of cancers. In addition, lncRNAs are reported to be involved in various steps of PC development and have a potential value in the diagnosis, treatment and prognostic prediction of PC. In this review, we highlight recent evidence related to the molecular mechanism of lncRNAs in growth, survival, invasion, metastasis, angiogenesis and apoptosis of PC cells, and discuss the potential clinical application of lncRNAs to the diagnosis, treatment and prognostic prediction of PC.
Collapse
Affiliation(s)
- Xiaoyi Huang
- Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xiaosong Zhi
- Department of Cell Biology, Second Military Medical University, Shanghai, China
| | - Yisha Gao
- Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Na Ta
- Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hui Jiang
- Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jianming Zheng
- Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
24
|
Del Castillo Velasco‐Herrera M, van der Weyden L, Nsengimana J, Speak AO, Sjöberg MK, Bishop DT, Jönsson G, Newton‐Bishop J, Adams DJ. Comparative genomics reveals that loss of lunatic fringe (LFNG) promotes melanoma metastasis. Mol Oncol 2018; 12:239-255. [PMID: 29193607 PMCID: PMC5792739 DOI: 10.1002/1878-0261.12161] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 10/16/2017] [Accepted: 11/07/2017] [Indexed: 12/17/2022] Open
Abstract
Metastasis is the leading cause of death in patients with advanced melanoma, yet the somatic alterations that aid tumour cell dissemination and colonisation are poorly understood. Here, we deploy comparative genomics to identify and validate clinically relevant drivers of melanoma metastasis. To do this, we identified a set of 976 genes whose expression level was associated with a poor outcome in patients from two large melanoma cohorts. Next, we characterised the genomes and transcriptomes of mouse melanoma cell lines defined as weakly metastatic, and their highly metastatic derivatives. By comparing expression data between species, we identified lunatic fringe (LFNG), among 28 genes whose expression level is predictive of poor prognosis and whose altered expression is associated with a prometastatic phenotype in mouse melanoma cells. CRISPR/Cas9-mediated knockout of Lfng dramatically enhanced the capability of weakly metastatic melanoma cells to metastasise in vivo, a phenotype that could be rescued with the Lfng cDNA. Notably, genomic alterations disrupting LFNG are found exclusively in human metastatic melanomas sequenced as part of The Cancer Genome Atlas. Using comparative genomics, we show that LFNG expression plays a functional role in regulating melanoma metastasis.
Collapse
Affiliation(s)
| | | | - Jeremie Nsengimana
- Leeds Institute of Cancer and PathologySt James's University HospitalUniversity of LeedsUK
| | - Anneliese O. Speak
- Experimental Cancer GeneticsWellcome Trust Sanger InstituteHinxtonCambridgeUK
| | - Marcela K. Sjöberg
- Experimental Cancer GeneticsWellcome Trust Sanger InstituteHinxtonCambridgeUK
- Departamento de Biología Celular y MolecularFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiagoChile
| | - David Timothy Bishop
- Leeds Institute of Cancer and PathologySt James's University HospitalUniversity of LeedsUK
| | - Göran Jönsson
- Division of Oncology and PathologyDepartment of Clinical SciencesSkåne University HospitalLund UniversitySweden
| | - Julia Newton‐Bishop
- Leeds Institute of Cancer and PathologySt James's University HospitalUniversity of LeedsUK
| | - David J. Adams
- Experimental Cancer GeneticsWellcome Trust Sanger InstituteHinxtonCambridgeUK
| |
Collapse
|
25
|
Harvey BM, Haltiwanger RS. Regulation of Notch Function by O-Glycosylation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1066:59-78. [PMID: 30030822 DOI: 10.1007/978-3-319-89512-3_4] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The Notch receptor initiates a unique intercellular signaling pathway that is evolutionarily conserved across all metazoans and contributes to the development and maintenance of numerous tissues. Consequently, many diseases result from aberrant Notch signaling. Emerging roles for Notch in disease are being uncovered as studies reveal new information regarding various components of this signaling pathway. Notch activity is regulated at several levels, but O-linked glycosylation of Epidermal Growth Factor (EGF) repeats in the Notch extracellular domain has emerged as a major regulator that, depending on context, can increase or decrease Notch activity. Three types of O-linked glycosylation occur at consensus sequences found within the EGF repeats of Notch: O-fucosylation, O-glucosylation, and O-GlcNAcylation. Recent studies have investigated the site occupancy of these types of glycosylation and also defined specific roles for these glycans on Notch structure and function. Nevertheless, there are many functional aspects to each type of O-glycosylation that remain unclear. Here, we will discuss molecular mechanisms of how O-glycosylation regulates Notch signaling and describe disorders associated with defects in Notch O-glycosylation.
Collapse
Affiliation(s)
- Beth M Harvey
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA.,Present Address: Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert S Haltiwanger
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA. .,Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA.
| |
Collapse
|
26
|
Zhang S, Liu J, Xu K, Li Z. Notch signaling via regulation of RB and p-AKT but not PIK3CG contributes to MIA PaCa-2 cell growth and migration to affect pancreatic carcinogenesis. Oncol Lett 2017; 15:2105-2110. [PMID: 29434912 PMCID: PMC5777124 DOI: 10.3892/ol.2017.7551] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 08/23/2017] [Indexed: 12/25/2022] Open
Abstract
Pancreatic cancer is one of the leading causes of cancer-associated mortality. The understanding of the expression pattern of key protein factors and their function in pancreatic cancer cells is therefore vital for the diagnosis and treatment of this malignancy. The results of the present study reveal that the levels of neurogenic locus notch homolog protein 2 (Notch2) and phosphorylated (p)-SMAD family member 2 decreased, whereas the expression of Notch3 and phosphoinositide-3 kinase catalytic subunit-γ protein increased in human pancreatic cancer tissues compared with tumor-adjacent tissues. Using the human pancreatic cancer MIA PaCa-2 cell line, it was observed that retinoblastoma-associated protein (RB) and p-RB expression were inhibited and p-AKT was upregulated when Notch signaling was activated in MIA PaCa-2 cells. Furthermore, inhibition of phosphoinositide-3 kinase catalytic subunit-γ (PIK3CG) activity by AS-605240 was able to block the growth and migration of MIA PaCa-2 cells. In conclusion, the results of the present study demonstrate that the Notch signal pathway may be involved in pancreatic carcinogenesis by modulating RB and p-AKT. PIK3CG may therefore be a potential target gene for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Shubing Zhang
- Department of Anesthesiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China.,Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, P.R. China
| | - Jingjiang Liu
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, P.R. China
| | - Keli Xu
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Zhijian Li
- Department of Anesthesiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
27
|
Cuomo D, Porreca I, Cobellis G, Tarallo R, Nassa G, Falco G, Nardone A, Rizzo F, Mallardo M, Ambrosino C. Carcinogenic risk and Bisphenol A exposure: A focus on molecular aspects in endoderm derived glands. Mol Cell Endocrinol 2017; 457:20-34. [PMID: 28111205 DOI: 10.1016/j.mce.2017.01.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 01/16/2017] [Accepted: 01/17/2017] [Indexed: 02/07/2023]
Abstract
Epidemiological and experimental evidence associates the exposure to Bisphenol A with the increase of cancer risk in several organs, including prostate. BPA targets different pathways involved in carcinogenicity including the Nuclear Receptors (i.e. estrogen and androgen receptors), stress regulated proteins and, finally, epigenetic changes. Here, we analyse BPA-dependent carcinogenesis in endoderm-derived glands, thyroid, liver, pancreas and prostate focusing on cell signalling, DNA damage repair pathways and epigenetic modifications. Mainly, we gather molecular data evidencing harmful effects at doses relevant for human risk (low-doses). Since few molecular data are available, above all for the pancreas, we analysed transcriptomic data generated in our laboratory to suggest possible mechanisms of BPA carcinogenicity in endoderm-derived glands, discussing the role of nuclear receptors and stress/NF-kB pathways. We evidence that an in vitro toxicogenomic approach might suggest mechanisms of toxicity applicable to cells having the same developmental origin. Although we cannot draw firm conclusions, published data summarized in this review suggest that exposure to BPA, primarily during the developmental stages, represents a risk for carcinogenesis of endoderm-derived glands.
Collapse
Affiliation(s)
- Danila Cuomo
- IRGS, Biogem, Via Camporeale, 83031 Ariano Irpino, Avellino, Italy; Department of Science and Technology, University of Sannio, via Port'Arsa 11, 82100 Benevento, Italy
| | | | - Gilda Cobellis
- Department of Experimental Medicine, Sez. Bozzatti, II University of Naples, 80138 Napoli, Italy
| | - Roberta Tarallo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Schola Medica Salernitana", University of Salerno, 84081 Baronissi, SA, Italy
| | - Giovanni Nassa
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Schola Medica Salernitana", University of Salerno, 84081 Baronissi, SA, Italy; Genomix4Life srl, Department of Medicine, Surgery and Dentistry "Schola Medica Salernitana", University of Salerno, Baronissi, SA, Italy
| | - Geppino Falco
- Department of Biology, University of Naples "Federico II", Napoli, Italy
| | - Antonio Nardone
- Department of Public Health, University of Naples "Federico II", Napoli, Italy
| | - Francesca Rizzo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Schola Medica Salernitana", University of Salerno, 84081 Baronissi, SA, Italy
| | - Massimo Mallardo
- Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Napoli, Italy
| | - Concetta Ambrosino
- Department of Science and Technology, University of Sannio, via Port'Arsa 11, 82100 Benevento, Italy.
| |
Collapse
|
28
|
Abstract
Cancer stem cells can generate tumors from only a small number of cells, whereas differentiated cancer cells cannot. The prominent feature of cancer stem cells is its ability to self-renew and differentiate into multiple types of cancer cells. Cancer stem cells have several distinct tumorigenic abilities, including stem cell signal transduction, tumorigenicity, metastasis, and resistance to anticancer drugs, which are regulated by genetic or epigenetic changes. Like normal adult stem cells involved in various developmental processes and tissue homeostasis, cancer stem cells maintain their self-renewal capacity by activating multiple stem cell signaling pathways and inhibiting differentiation signaling pathways during cancer initiation and progression. Recently, many studies have focused on targeting cancer stem cells to eradicate malignancies by regulating stem cell signaling pathways, and products of some of these strategies are in preclinical and clinical trials. In this review, we describe the crucial features of cancer stem cells related to tumor relapse and drug resistance, as well as the new therapeutic strategy to target cancer stem cells named "differentiation therapy."
Collapse
Affiliation(s)
- Xiong Jin
- 1 Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
- 2 Institute of Animal Molecular Biotechnology, Korea University, Seoul, Republic of Korea
| | - Xun Jin
- 3 Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- 4 Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- 5 Institute of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hyunggee Kim
- 1 Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
- 2 Institute of Animal Molecular Biotechnology, Korea University, Seoul, Republic of Korea
| |
Collapse
|
29
|
Kras G12D upregulates Notch signaling to induce gallbladder tumorigenesis in mice. Oncoscience 2017; 4:131-138. [PMID: 29142904 PMCID: PMC5672897 DOI: 10.18632/oncoscience.368] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 09/17/2017] [Indexed: 11/25/2022] Open
Abstract
Background Kras mutations and increased Notch activation occur frequently in gallbladder cancer. However, their roles in gallbladder carcinogenesis have not been defined. This study was aimed at determining whether expression of mutant Kras was sufficient to induce gallbladder carcinoma and whether Notch deregulation played a role in this context. Methods We determined Cre recombination activity of Pdx1-Cre in the gallbladder using a reporter strain and examined gallbladder tumor development in the KrasLSL- G12D/+;Pdx1-Cre mice. We analyzed expression of Notch pathway genes in the mouse gallbladder by immunohistochemistry, quantitative RT-PCR, and Western blot analysis. We also determined the effect of Jag1 deletion on Kras-induced gallbladder tumor development. Results Pdx1-Cre exhibits robust recombination activity in the gallbladder epithelium. KrasLSL-G12D/+;Pdx1-Cre mice form early onset adenoma in the gallbladder and adjacent biliary tract with complete penetrance, albeit short of invasive adenocarcinoma. KrasG12D upregulates expressions of Notch2, Notch3, Notch4, Jag1 and downstream target genes Hes1, Hey1 and Hey2, and deletion of Jag1 partially suppresses KrasG12D-induced adenoma development. Conclusions KrasG12D induces gallbladder adenoma and Notch plays a key role in Kras-initiated gallbladder tumorigenesis.
Collapse
|
30
|
Gao J, Long B, Wang Z. Role of Notch signaling pathway in pancreatic cancer. Am J Cancer Res 2017; 7:173-186. [PMID: 28337369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 10/12/2016] [Indexed: 09/28/2022] Open
Abstract
Pancreatic cancer (PC) is one of the highly aggressive malignancies in the United States. It has been shown that multiple signaling pathways are involved in the pathogenesis of PC, such as JNK, PI3K/AKT, Rho GTPase, Hedgehog (Hh) and Skp2. In recent years, accumulated evidence has demonstrated that Notch signaling pathway plays critical roles in the development and progression of PC. Therefore, in this review we discuss the recent literature regarding the function and regulation of Notch in the pathogenesis of PC. Moreover, we describe that Notch signaling pathway could be down-regulated by its inhibitors or natural compounds, which could be a novel approach for the treatment of PC patients.
Collapse
Affiliation(s)
- Jiankun Gao
- Sichuan College of Tranditional Chinese Medicine Mianyang, Sichuan, China
| | - Bo Long
- Department of Infectious Diseases, Mianyang 404 Hospital Mianyang, Sichuan, China
| | - Zhiwei Wang
- The Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital, Soochow UniversitySuzhou 215123, China; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical SchoolMA 02215, USA
| |
Collapse
|
31
|
Módos D, Bulusu KC, Fazekas D, Kubisch J, Brooks J, Marczell I, Szabó PM, Vellai T, Csermely P, Lenti K, Bender A, Korcsmáros T. Neighbours of cancer-related proteins have key influence on pathogenesis and could increase the drug target space for anticancer therapies. NPJ Syst Biol Appl 2017; 3:2. [PMID: 28603644 PMCID: PMC5460138 DOI: 10.1038/s41540-017-0003-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Even targeted chemotherapies against solid cancers show a moderate success increasing the need to novel targeting strategies. To address this problem, we designed a systems-level approach investigating the neighbourhood of mutated or differentially expressed cancer-related proteins in four major solid cancers (colon, breast, liver and lung). Using signalling and protein–protein interaction network resources integrated with mutational and expression datasets, we analysed the properties of the direct and indirect interactors (first and second neighbours) of cancer-related proteins, not found previously related to the given cancer type. We found that first neighbours have at least as high degree, betweenness centrality and clustering coefficient as cancer-related proteins themselves, indicating a previously unknown central network position. We identified a complementary strategy for mutated and differentially expressed proteins, where the affect of differentially expressed proteins having smaller network centrality is compensated with high centrality first neighbours. These first neighbours can be considered as key, so far hidden, components in cancer rewiring, with similar importance as mutated proteins. These observations strikingly suggest targeting first neighbours as a novel strategy for disrupting cancer-specific networks. Remarkably, our survey revealed 223 marketed drugs already targeting first neighbour proteins but applied mostly outside oncology, providing a potential list for drug repurposing against solid cancers. For the very central first neighbours, whose direct targeting would cause several side effects, we suggest a cancer-mimicking strategy by targeting their interactors (second neighbours of cancer-related proteins, having a central protein affecting position, similarly to the cancer-related proteins). Hence, we propose to include first neighbours to network medicine based approaches for (but not limited to) anticancer therapies. Cancer is considered a systems disease in which the interactors of cancer-related proteins have a key role, also as targets to fight cancer. New therapeutic approaches are needed to improve success rates and to identify suitable proteins as novel, alternative drug targets. We designed a computational approach, combining mutation and differential expression data with network information, to analyse the interactions of cancer-related proteins in colon, breast, liver and lung cancer. We found that first (direct) neighbours, not linked previously to the given cancer type, are similarly important as mutated proteins known to be involved in cancer development. We found 223 drugs already in the clinic targeting these proteins but not yet used against cancer as their oncology relevance was hidden so far. Our observations open up new strategies for target selection and anti-cancer drug discovery.
Collapse
Affiliation(s)
- Dezső Módos
- Department of Morphology and Physiology, Department of Health Science, Semmelweis University, Budapest, Hungary.,Department of Genetics, Eötvös Loránd University, Budapest, Hungary.,Earlham Institute, Norwich Research Park, Norwich, UK.,Gut Health and Food Safety Programme, Institute of Food Research, Norwich Research Park, Norwich, UK
| | - Krishna C Bulusu
- Centre for Molecular Informatics, University of Cambridge, Cambridge, UK
| | - Dávid Fazekas
- Department of Genetics, Eötvös Loránd University, Budapest, Hungary.,Gut Health and Food Safety Programme, Institute of Food Research, Norwich Research Park, Norwich, UK
| | - János Kubisch
- Department of Genetics, Eötvös Loránd University, Budapest, Hungary
| | - Johanne Brooks
- Gut Health and Food Safety Programme, Institute of Food Research, Norwich Research Park, Norwich, UK.,Department of Medicine and Health, University of East Anglia, Norwich, UK.,Department of Gastroenterology, Norfolk and Norwich University Hospitals, Norwich, UK
| | - István Marczell
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Péter M Szabó
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary.,Biometric Research Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tibor Vellai
- Department of Genetics, Eötvös Loránd University, Budapest, Hungary
| | - Péter Csermely
- Department of Medical Chemistry, Semmelweis University, Budapest, Hungary
| | - Katalin Lenti
- Department of Morphology and Physiology, Department of Health Science, Semmelweis University, Budapest, Hungary
| | - Andreas Bender
- Centre for Molecular Informatics, University of Cambridge, Cambridge, UK
| | - Tamás Korcsmáros
- Department of Genetics, Eötvös Loránd University, Budapest, Hungary.,Earlham Institute, Norwich Research Park, Norwich, UK.,Gut Health and Food Safety Programme, Institute of Food Research, Norwich Research Park, Norwich, UK
| |
Collapse
|
32
|
Quantitative secretomic analysis of pancreatic cancer cells in serum-containing conditioned medium. Sci Rep 2016; 6:37606. [PMID: 27869176 PMCID: PMC5116583 DOI: 10.1038/srep37606] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 11/01/2016] [Indexed: 01/22/2023] Open
Abstract
Pancreatic cancer is a highly metastatic and chemo-resistant disease. Secreted proteins involved in cell-cell interactions play an important role in changing the tumor microenvironment. Previous studies generally focus on the secretome of cancer cell line from serum-free media, due to the serious interference of fetal bovine serum (FBS). However, serum-starvation may alter expression patterns of secreted proteins. Hence, efforts to decrease the interference of serum in proteomic analysis of serum-containing media have been hampered to quantitatively measure the tumor secretion levels. Recently, the metabolic labeling, protein equalization, protein fractionation and filter-aided sample preparation (FASP) strategy (MLEFF) has been successfully used to avoid the disturbance of serum on secretome analysis. Here, this efficient method was applied for comparative secretome analysis of two hamster pancreatic cancer cells with differentially metastatic potentials, enabling the observation of 161 differentially expressed proteins, including 106 proteins that had been previously reported and detected in plasma. By integrated analysis of our data and publicly available bioinformatics resources, we found that a combination panel consisting of CDH3, PLAU, and LFNG might improve the prognosis of overall pancreatic cancer survival. These secreted proteins may serve as a potential therapeutic targets for pancreatic cancer metastasis.
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW The review intends to describe recent studies on the development of pancreatic cancer from a genetic, molecular, and microenvironment perspective. RECENT FINDINGS Pancreatic cancer has been discovered to have distinct molecular subtypes based on transcriptome analyses that may have implications for treatment. Recent studies are also mapping the complex molecular biology of this cancer as it relates to the core signaling abnormalities inherent to this disease. There have been discoveries of novel modes of regulation of pancreatic cancer development, including alterations in key transcription factors, epigenetic modifiers, and metabolic pathways. Studies of the tumor-associated microenvironment continue to reveal its complex role in tumor development. SUMMARY Pancreatic cancer development appears to depend on a multifaceted network of signals that are dynamic, involve multiple cell types, and are linked to spatiotemporal factors in tumor evolution. Understanding the development of pancreatic cancer in this context is key to identifying novel and effective targets for treatment.
Collapse
|